1
|
Han TW, Portz B, Young RA, Boija A, Klein IA. RNA and condensates: Disease implications and therapeutic opportunities. Cell Chem Biol 2024; 31:1593-1609. [PMID: 39303698 DOI: 10.1016/j.chembiol.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024]
Abstract
Biomolecular condensates are dynamic membraneless organelles that compartmentalize proteins and RNA molecules to regulate key cellular processes. Diverse RNA species exert their effects on the cell by their roles in condensate formation and function. RNA abnormalities such as overexpression, modification, and mislocalization can lead to pathological condensate behaviors that drive various diseases, including cancer, neurological disorders, and infections. Here, we review RNA's role in condensate biology, describe the mechanisms of RNA-induced condensate dysregulation, note the implications for disease pathogenesis, and discuss novel therapeutic strategies. Emerging approaches to targeting RNA within condensates, including small molecules and RNA-based therapies that leverage the unique properties of condensates, may revolutionize treatment for complex diseases.
Collapse
Affiliation(s)
| | | | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ann Boija
- Dewpoint Therapeutics, Boston, MA, USA.
| | | |
Collapse
|
2
|
Ruan K, Bai G, Fang Y, Li D, Li T, Liu X, Lu B, Lu Q, Songyang Z, Sun S, Wang Z, Zhang X, Zhou W, Zhang H. Biomolecular condensates and disease pathogenesis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1792-1832. [PMID: 39037698 DOI: 10.1007/s11427-024-2661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Biomolecular condensates or membraneless organelles (MLOs) formed by liquid-liquid phase separation (LLPS) divide intracellular spaces into discrete compartments for specific functions. Dysregulation of LLPS or aberrant phase transition that disturbs the formation or material states of MLOs is closely correlated with neurodegeneration, tumorigenesis, and many other pathological processes. Herein, we summarize the recent progress in development of methods to monitor phase separation and we discuss the biogenesis and function of MLOs formed through phase separation. We then present emerging proof-of-concept examples regarding the disruption of phase separation homeostasis in a diverse array of clinical conditions including neurodegenerative disorders, hearing loss, cancers, and immunological diseases. Finally, we describe the emerging discovery of chemical modulators of phase separation.
Collapse
Affiliation(s)
- Ke Ruan
- The First Affiliated Hospital & School of Life Sciences, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Ge Bai
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Tingting Li
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| | - Qing Lu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Zhou Songyang
- State Key Laboratory of Biocontrol, MOE Key Laboratory of Gene Function and Regulation and Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Shuguo Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zheng Wang
- The Second Affiliated Hospital, School of Basic Medical Sciences, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Xin Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
| | - Wen Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Hong Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
3
|
Felicetti T, Sarnari C, Gaito R, Tabarrini O, Manfroni G. Recent Progress toward the Discovery of Small Molecules as Novel Anti-Respiratory Syncytial Virus Agents. J Med Chem 2024; 67:11543-11579. [PMID: 38970494 DOI: 10.1021/acs.jmedchem.4c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Respiratory syncytial virus (RSV) stands as the foremost cause of infant hospitalization globally, ranking second only to malaria in terms of infant mortality. Although three vaccines have recently been approved for the prophylaxis of adults aged 60 and above, and pregnant women, there is currently no effective antiviral drug for treating RSV infections. The only preventive measure for infants at high risk of severe RSV disease is passive immunization through monoclonal antibodies. This Perspective offers an overview of the latest advancements in RSV drug discovery of small molecule antivirals, with particular focus on the promising findings from agents targeting the fusion and polymerase proteins. A comprehensive reflection on the current state of RSV research is also given, drawing inspiration from the lessons gleaned from HCV and HIV, while also considering the impact of the recent approval of the three vaccines.
Collapse
Affiliation(s)
- Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Chiara Sarnari
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Roberta Gaito
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| |
Collapse
|
4
|
Yang Q, Xue B, Liu F, Lu Y, Tang J, Yan M, Wu Q, Chen R, Zhou A, Liu L, Liu J, Qu C, Wu Q, Fu M, Zhong J, Dong J, Chen S, Wang F, Zhou Y, Zheng J, Peng W, Shang J, Chen X. Farnesyltransferase inhibitor lonafarnib suppresses respiratory syncytial virus infection by blocking conformational change of fusion glycoprotein. Signal Transduct Target Ther 2024; 9:144. [PMID: 38853183 PMCID: PMC11163014 DOI: 10.1038/s41392-024-01858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/28/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the major cause of bronchiolitis and pneumonia in young children and the elderly. There are currently no approved RSV-specific therapeutic small molecules available. Using high-throughput antiviral screening, we identified an oral drug, the prenylation inhibitor lonafarnib, which showed potent inhibition of the RSV fusion process. Lonafarnib exhibited antiviral activity against both the RSV A and B genotypes and showed low cytotoxicity in HEp-2 and human primary bronchial epithelial cells (HBEC). Time-of-addition and pseudovirus assays demonstrated that lonafarnib inhibits RSV entry, but has farnesyltransferase-independent antiviral efficacy. Cryo-electron microscopy revealed that lonafarnib binds to a triple-symmetric pocket within the central cavity of the RSV F metastable pre-fusion conformation. Mutants at the RSV F sites interacting with lonafarnib showed resistance to lonafarnib but remained fully sensitive to the neutralizing monoclonal antibody palivizumab. Furthermore, lonafarnib dose-dependently reduced the replication of RSV in BALB/c mice. Collectively, lonafarnib could be a potential fusion inhibitor for RSV infection.
Collapse
Affiliation(s)
- Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fengjiang Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Lu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengrong Yan
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiong Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ruyi Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Anqi Zhou
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Lijie Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junjun Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Changbin Qu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Qingxin Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Muqing Fu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jiayi Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jianwei Dong
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Sijie Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Fan Wang
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuan Zhou
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou lnstitute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Wei Peng
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jinsai Shang
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
5
|
Zhang Q, Ye H, Liu C, Zhou H, He M, Liang X, Zhou Y, Wang K, Qin Y, Li Z, Chen M. PABP-driven secondary condensed phase within RSV inclusion bodies activates viral mRNAs for ribosomal recruitment. Virol Sin 2024; 39:235-250. [PMID: 38072230 PMCID: PMC11074649 DOI: 10.1016/j.virs.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/03/2023] [Indexed: 01/12/2024] Open
Abstract
Inclusion bodies (IBs) of respiratory syncytial virus (RSV) are formed by liquid-liquid phase separation (LLPS) and contain internal structures termed "IB-associated granules" (IBAGs), where anti-termination factor M2-1 and viral mRNAs are concentrated. However, the mechanism of IBAG formation and the physiological function of IBAGs are unclear. Here, we found that the internal structures of RSV IBs are actual M2-1-free viral messenger ribonucleoprotein (mRNP) condensates formed by secondary LLPS. Mechanistically, the RSV nucleoprotein (N) and M2-1 interact with and recruit PABP to IBs, promoting PABP to bind viral mRNAs transcribed in IBs by RNA-recognition motif and drive secondary phase separation. Furthermore, PABP-eIF4G1 interaction regulates viral mRNP condensate composition, thereby recruiting specific translation initiation factors (eIF4G1, eIF4E, eIF4A, eIF4B and eIF4H) into the secondary condensed phase to activate viral mRNAs for ribosomal recruitment. Our study proposes a novel LLPS-regulated translation mechanism during viral infection and a novel antiviral strategy via targeting on secondary condensed phase.
Collapse
Affiliation(s)
- Qiang Zhang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Hanzhe Ye
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Cong Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Haiwu Zhou
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Mingbin He
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiaodong Liang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yu Zhou
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Kun Wang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yali Qin
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Zhifei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Mingzhou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| |
Collapse
|
6
|
Martin EW, Iserman C, Olety B, Mitrea DM, Klein IA. Biomolecular Condensates as Novel Antiviral Targets. J Mol Biol 2024; 436:168380. [PMID: 38061626 DOI: 10.1016/j.jmb.2023.168380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
Viral infections pose a significant health risk worldwide. There is a pressing need for more effective antiviral drugs to combat emerging novel viruses and the reemergence of previously controlled viruses. Biomolecular condensates are crucial for viral replication and are promising targets for novel antiviral therapies. Herein, we review the role of biomolecular condensates in the viral replication cycle and discuss novel strategies to leverage condensate biology for antiviral drug discovery. Biomolecular condensates may also provide an opportunity to develop antivirals that are broad-spectrum or less prone to acquired drug resistance.
Collapse
|
7
|
Langedijk AC, Bont LJ. Respiratory syncytial virus infection and novel interventions. Nat Rev Microbiol 2023; 21:734-749. [PMID: 37438492 DOI: 10.1038/s41579-023-00919-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/14/2023]
Abstract
The large global burden of respiratory syncytial virus (RSV) respiratory tract infections in young children and older adults has gained increased recognition in recent years. Recent discoveries regarding the neutralization-specific viral epitopes of the pre-fusion RSV glycoprotein have led to a shift from empirical to structure-based design of RSV therapeutics, and controlled human infection model studies have provided early-stage proof of concept for novel RSV monoclonal antibodies, vaccines and antiviral drugs. The world's first vaccines and first monoclonal antibody to prevent RSV among older adults and all infants, respectively, have recently been approved. Large-scale introduction of RSV prophylactics emphasizes the need for active surveillance to understand the global impact of these interventions over time and to timely identify viral mutants that are able to escape novel prophylactics. In this Review, we provide an overview of RSV interventions in clinical development, highlighting global disease burden, seasonality, pathogenesis, and host and viral factors related to RSV immunity.
Collapse
Affiliation(s)
- Annefleur C Langedijk
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Louis J Bont
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands.
- ReSViNET Foundation, Zeist, the Netherlands.
| |
Collapse
|
8
|
Jahajeeah D, Ranghoo-Sanmukhiya M, Schäfer G. Metabolic Profiling, Antiviral Activity and the Microbiome of Some Mauritian Soft Corals. Mar Drugs 2023; 21:574. [PMID: 37999398 PMCID: PMC10672535 DOI: 10.3390/md21110574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
Soft corals, recognized as sessile marine invertebrates, rely mainly on chemical, rather than physical defense, by secreting intricate secondary metabolites with plausible pharmaceutical implication. Their ecological niche encompasses a diverse community of symbiotic microorganisms which potentially contribute to the biosynthesis of these bioactive metabolites. The emergence of new viruses and heightened viral resistance underscores the urgency to explore novel pharmacological reservoirs. Thus, marine organisms, notably soft corals and their symbionts, have drawn substantial attention. In this study, the chemical composition of four Mauritian soft corals: Sinularia polydactya, Cespitularia simplex, Lobophytum patulum, and Lobophytum crassum was investigated using LC-MS techniques. Concurrently, Illumina 16S metagenomic sequencing was used to identify the associated bacterial communities in the named soft corals. The presence of unique biologically important compounds and vast microbial communities found therein was further followed up to assess their antiviral effects against SARS-CoV-2 and HPV pseudovirus infection. Strikingly, among the studied soft corals, L. patulum displayed an expansive repertoire of unique metabolites alongside a heightened bacterial consort. Moreover, L. patulum extracts exerted some promising antiviral activity against SARS-CoV-2 and HPV pseudovirus infection, and our findings suggest that L. patulum may have the potential to serve as a therapeutic agent in the prevention of infectious diseases, thereby warranting further investigation.
Collapse
Affiliation(s)
- Deeya Jahajeeah
- Department of Agricultural & Food Science, Faculty of Agriculture, University of Mauritius, Reduit 80837, Mauritius;
- International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa;
| | - Mala Ranghoo-Sanmukhiya
- Department of Agricultural & Food Science, Faculty of Agriculture, University of Mauritius, Reduit 80837, Mauritius;
| | - Georgia Schäfer
- International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa;
| |
Collapse
|
9
|
Abstract
Biomolecular condensates formed by phase separation are widespread and play critical roles in many physiological and pathological processes. cGAS-STING signaling functions to detect aberrant DNA signals to initiate anti-infection defense and antitumor immunity. At the same time, cGAS-STING signaling must be carefully regulated to maintain immune homeostasis. Interestingly, exciting recent studies have reported that biomolecular phase separation exists and plays important roles in different steps of cGAS-STING signaling, including cGAS condensates, STING condensates, and IRF3 condensates. In addition, several intracellular and extracellular factors have been proposed to modulate the condensates in cGAS-STING signaling. These studies reveal novel activation and regulation mechanisms of cGAS-STING signaling and provide new opportunities for drug discovery. Here, we summarize recent advances in the phase separation of cGAS-STING signaling and the development of potential drugs targeting these innate immune condensates.
Collapse
Affiliation(s)
- Quanjin Li
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Pu Gao
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
10
|
Diot C, Richard CA, Risso-Ballester J, Martin D, Fix J, Eléouët JF, Sizun C, Rameix-Welti MA, Galloux M. Hardening of Respiratory Syncytial Virus Inclusion Bodies by Cyclopamine Proceeds through Perturbation of the Interactions of the M2-1 Protein with RNA and the P Protein. Int J Mol Sci 2023; 24:13862. [PMID: 37762166 PMCID: PMC10531356 DOI: 10.3390/ijms241813862] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/20/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Respiratory syncytial virus (RSV) RNA synthesis takes place in cytoplasmic viral factories also called inclusion bodies (IBs), which are membrane-less organelles concentrating the viral RNA polymerase complex. The assembly of IBs is driven by liquid-liquid phase separation promoted by interactions between the viral nucleoprotein N and the phosphoprotein P. We recently demonstrated that cyclopamine (CPM) inhibits RSV multiplication by disorganizing and hardening IBs. Although a single mutation in the viral transcription factor M2-1 induced resistance to CPM, the mechanism of action of CPM still remains to be characterized. Here, using FRAP experiments on reconstituted pseudo-IBs both in cellula and in vitro, we first demonstrated that CPM activity depends on the presence of M2-1 together with N and P. We showed that CPM impairs the competition between P and RNA binding to M2-1. As mutations on both P and M2-1 induced resistance against CPM activity, we suggest that CPM may affect the dynamics of the M2-1-P interaction, thereby affecting the relative mobility of the proteins contained in RSV IBs. Overall, our results reveal that stabilizing viral protein-protein interactions is an attractive new antiviral approach. They pave the way for the rational chemical optimization of new specific anti-RSV molecules.
Collapse
Affiliation(s)
- Cédric Diot
- Institut Pasteur, Université Paris Cité, M3P, F-75015 Paris, France;
- INSERM, UMR 1173 (2I), Université Paris-Saclay-Versailles St. Quentin, M3P, F-78180 Versailles, France;
| | - Charles-Adrien Richard
- INRAE, Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St. Quentin, F-78350 Jouy-en-Josas, France; (C.-A.R.); (D.M.); (J.F.); (J.-F.E.)
| | - Jennifer Risso-Ballester
- INSERM, UMR 1173 (2I), Université Paris-Saclay-Versailles St. Quentin, M3P, F-78180 Versailles, France;
| | - Davy Martin
- INRAE, Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St. Quentin, F-78350 Jouy-en-Josas, France; (C.-A.R.); (D.M.); (J.F.); (J.-F.E.)
| | - Jenna Fix
- INRAE, Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St. Quentin, F-78350 Jouy-en-Josas, France; (C.-A.R.); (D.M.); (J.F.); (J.-F.E.)
| | - Jean-François Eléouët
- INRAE, Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St. Quentin, F-78350 Jouy-en-Josas, France; (C.-A.R.); (D.M.); (J.F.); (J.-F.E.)
| | - Christina Sizun
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, F-91190 Gif-sur-Yvette, France;
| | - Marie-Anne Rameix-Welti
- INSERM, UMR 1173 (2I), Université Paris-Saclay-Versailles St. Quentin, M3P, F-78180 Versailles, France;
- Laboratoire de Microbiologie, Hôpital Ambroise Paré, Assistance Publique des Hôpitaux de Paris, DMU15, F-75015 Paris, France
| | - Marie Galloux
- INRAE, Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St. Quentin, F-78350 Jouy-en-Josas, France; (C.-A.R.); (D.M.); (J.F.); (J.-F.E.)
| |
Collapse
|
11
|
Li S, Wang Y, Lai L. Small molecules in regulating protein phase separation. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1075-1083. [PMID: 37294104 PMCID: PMC10415206 DOI: 10.3724/abbs.2023106] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/15/2023] [Indexed: 06/10/2023] Open
Abstract
Biomolecular condensates formed by phase separation are involved in many cellular processes. Dysfunctional or abnormal condensates are closely associated with neurodegenerative diseases, cancer and other diseases. Small molecules can effectively regulate protein phase separation by modulating the formation, dissociation, size and material properties of condensates. Discovery of small molecules to regulate protein phase separation provides chemical probes for deciphering the underlying mechanism and potential novel treatments for condensate-related diseases. Here we review the advances of small molecule regulation of phase separation. The discovery, chemical structures of recently found small molecule phase separation regulators and how they modulate biological condensates are summarized and discussed. Possible strategies to accelerate the discovery of more liquid-liquid phase separation (LLPS)-regulating small molecules are proposed.
Collapse
Affiliation(s)
- Siyang Li
- Center for Quantitative BiologyAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Yanyan Wang
- BNLMSPeking-Tsinghua Center for Life Sciences at College of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| | - Luhua Lai
- Center for Quantitative BiologyAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
- BNLMSPeking-Tsinghua Center for Life Sciences at College of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| |
Collapse
|
12
|
Visentin A, Demitroff N, Salgueiro M, Borkosky SS, Uversky VN, Camporeale G, de Prat-Gay G. Assembly of the Tripartite and RNA Condensates of the Respiratory Syncytial Virus Factory Proteins In Vitro: Role of the Transcription Antiterminator M 2-1. Viruses 2023; 15:1329. [PMID: 37376628 DOI: 10.3390/v15061329] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
A wide variety of viruses replicate in liquid-like viral factories. Non-segmented negative stranded RNA viruses share a nucleoprotein (N) and a phosphoprotein (P) that together emerge as the main drivers of liquid-liquid phase separation. The respiratory syncytial virus includes the transcription antiterminator M2-1, which binds RNA and maximizes RNA transcriptase processivity. We recapitulate the assembly mechanism of condensates of the three proteins and the role played by RNA. M2-1 displays a strong propensity for condensation by itself and with RNA through the formation of electrostatically driven protein-RNA coacervates based on the amphiphilic behavior of M2-1 and finely tuned by stoichiometry. M2-1 incorporates into tripartite condensates with N and P, modulating their size through an interplay with P, where M2-1 is both client and modulator. RNA is incorporated into the tripartite condensates adopting a heterogeneous distribution, reminiscent of the M2-1-RNA IBAG granules within the viral factories. Ionic strength dependence indicates that M2-1 behaves differently in the protein phase as opposed to the protein-RNA phase, in line with the subcompartmentalization observed in viral factories. This work dissects the biochemical grounds for the formation and fate of the RSV condensates in vitro and provides clues to interrogate the mechanism under the highly complex infection context.
Collapse
Affiliation(s)
- Araceli Visentin
- Instituto Leloir, IIB-BA Conicet, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Nicolás Demitroff
- Instituto Leloir, IIB-BA Conicet, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Mariano Salgueiro
- Instituto Leloir, IIB-BA Conicet, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Silvia Susana Borkosky
- Instituto Leloir, IIB-BA Conicet, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Gabriela Camporeale
- Instituto Leloir, IIB-BA Conicet, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Gonzalo de Prat-Gay
- Instituto Leloir, IIB-BA Conicet, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-970, SP, Brazil
| |
Collapse
|
13
|
Fix J, Descamps D, Galloux M, Ferret C, Bouguyon E, Zohari S, Näslund K, Hägglund S, Altmeyer R, Valarcher JF, Riffault S, Eléouët JF. Screening antivirals with a mCherry-expressing recombinant bovine respiratory syncytial virus: a proof of concept using cyclopamine. Vet Res 2023; 54:36. [PMID: 37069656 PMCID: PMC10111787 DOI: 10.1186/s13567-023-01165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 03/17/2023] [Indexed: 04/19/2023] Open
Abstract
Bovine respiratory syncytial virus (BRSV) is a pathogenic pneumovirus and a major cause of acute respiratory infections in calves. Although different vaccines are available against BRSV, their efficiency remains limited, and no efficient and large-scale treatment exists. Here, we developed a new reverse genetics system for BRSV expressing the red fluorescent protein mCherry, based on a field strain isolated from a sick calf in Sweden. Although this recombinant fluorescent virus replicated slightly less efficiently compared to the wild type virus, both viruses were shown to be sensitive to the natural steroidal alkaloid cyclopamine, which was previously shown to inhibit human RSV replication. Our data thus point to the potential of this recombinant fluorescent BRSV as a powerful tool in preclinical drug discovery to enable high throughput compound screening.
Collapse
Affiliation(s)
- Jenna Fix
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Delphyne Descamps
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Marie Galloux
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Cécile Ferret
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Edwige Bouguyon
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Siamak Zohari
- Department of Microbiology, National Veterinary Institute (SVA), Uppsala, Sweden
| | - Katarina Näslund
- Department of Microbiology, National Veterinary Institute (SVA), Uppsala, Sweden
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sara Hägglund
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Jean-François Valarcher
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sabine Riffault
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
14
|
Ponticelli M, Bellone ML, Parisi V, Iannuzzi A, Braca A, de Tommasi N, Russo D, Sileo A, Quaranta P, Freer G, Pistello M, Milella L. Specialized metabolites from plants as a source of new multi-target antiviral drugs: a systematic review. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2023; 22:1-79. [PMID: 37359711 PMCID: PMC10008214 DOI: 10.1007/s11101-023-09855-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/30/2023] [Indexed: 06/28/2023]
Abstract
Viral infections have always been the main global health challenge, as several potentially lethal viruses, including the hepatitis virus, herpes virus, and influenza virus, have affected human health for decades. Unfortunately, most licensed antiviral drugs are characterized by many adverse reactions and, in the long-term therapy, also develop viral resistance; for these reasons, researchers have focused their attention on investigating potential antiviral molecules from plants. Natural resources indeed offer a variety of specialized therapeutic metabolites that have been demonstrated to inhibit viral entry into the host cells and replication through the regulation of viral absorption, cell receptor binding, and competition for the activation of intracellular signaling pathways. Many active phytochemicals, including flavonoids, lignans, terpenoids, coumarins, saponins, alkaloids, etc., have been identified as potential candidates for preventing and treating viral infections. Using a systematic approach, this review summarises the knowledge obtained to date on the in vivo antiviral activity of specialized metabolites extracted from plant matrices by focusing on their mechanism of action.
Collapse
Affiliation(s)
- Maria Ponticelli
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| | - Maria Laura Bellone
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
- Ph.D. Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Valentina Parisi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
- Ph.D. Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Annamaria Iannuzzi
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126 Pisa, Italy
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandra Braca
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126 Pisa, Italy
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Nunziatina de Tommasi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Daniela Russo
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| | - Annalisa Sileo
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| | | | - Giulia Freer
- Virology Unit, Pisa University Hospital, Pisa, Italy
| | | | - Luigi Milella
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
15
|
Sutto-Ortiz P, Eléouët JF, Ferron F, Decroly E. Biochemistry of the Respiratory Syncytial Virus L Protein Embedding RNA Polymerase and Capping Activities. Viruses 2023; 15:v15020341. [PMID: 36851554 PMCID: PMC9960070 DOI: 10.3390/v15020341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/12/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
The human respiratory syncytial virus (RSV) is a negative-sense, single-stranded RNA virus. It is the major cause of severe acute lower respiratory tract infection in infants, the elderly population, and immunocompromised individuals. There is still no approved vaccine or antiviral treatment against RSV disease, but new monoclonal prophylactic antibodies are yet to be commercialized, and clinical trials are in progress. Hence, urgent efforts are needed to develop efficient therapeutic treatments. RSV RNA synthesis comprises viral transcription and replication that are catalyzed by the large protein (L) in coordination with the phosphoprotein polymerase cofactor (P), the nucleoprotein (N), and the M2-1 transcription factor. The replication/transcription is orchestrated by the L protein, which contains three conserved enzymatic domains: the RNA-dependent RNA polymerase (RdRp), the polyribonucleotidyl transferase (PRNTase or capping), and the methyltransferase (MTase) domain. These activities are essential for the RSV replicative cycle and are thus considered as attractive targets for the development of therapeutic agents. In this review, we summarize recent findings about RSV L domains structure that highlight how the enzymatic activities of RSV L domains are interconnected, discuss the most relevant and recent antivirals developments that target the replication/transcription complex, and conclude with a perspective on identified knowledge gaps that enable new research directions.
Collapse
Affiliation(s)
| | - Jean-François Eléouët
- Unité de Virologie et Immunologie Moléculaires, INRAE, Université Paris Saclay, F78350 Jouy en Josas, France
| | - François Ferron
- Aix Marseille Université, CNRS, AFMB, UMR, 7257 Marseille, France
- European Virus Bioinformatics Center, Leutragraben 1, 07743 Jena, Germany
| | - Etienne Decroly
- Aix Marseille Université, CNRS, AFMB, UMR, 7257 Marseille, France
- Correspondence:
| |
Collapse
|
16
|
Phytocompounds as a source for the development of new drugs to treat respiratory viral infections. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2023; 77:187-240. [PMCID: PMC10204935 DOI: 10.1016/b978-0-323-91294-5.00007-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
Respiratory viruses have an important history as a threat to global health. However, this problem has been aggravated due to the appearance of new outbreaks caused by a newly discovered virus or variant. Recently, the new coronavirus (SARS-CoV-2) has been a major concern for health authorities, and it was classified as a pandemic by the World Health Organization. Secondary metabolites obtained from plants represent an alternative to the discovery of new active molecules and have already shown potential to combat different viruses. In an effort to demonstrate the broad spectrum of antiviral action from these metabolites, this work describes the compounds that were effective against the major viruses that cause respiratory infections in humans. In addition, their mechanisms of action were highlighted as an approach to better understanding the virus-bioactive substance relationship. Finally, this study warns that, although phytocompounds have a broad antiviral action spectrum, the development of products and clinical trials based on these secondary metabolites is still scarce and therefore deserves greater attention from the scientific community.
Collapse
|
17
|
Sourimant J, Lieber CM, Yoon JJ, Toots M, Govindarajan M, Udumula V, Sakamoto K, Natchus MG, Patti J, Vernachio J, Plemper RK. Orally efficacious lead of the AVG inhibitor series targeting a dynamic interface in the respiratory syncytial virus polymerase. SCIENCE ADVANCES 2022; 8:eabo2236. [PMID: 35749502 PMCID: PMC9232112 DOI: 10.1126/sciadv.abo2236] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory infections in infants and the immunocompromised, yet no efficient therapeutic exists. We have identified the AVG class of allosteric inhibitors of RSV RNA synthesis. Here, we demonstrate through biolayer interferometry and in vitro RNA-dependent RNA polymerase (RdRP) assays that AVG compounds bind to the viral polymerase, stalling the polymerase in initiation conformation. Resistance profiling revealed a unique escape pattern, suggesting a discrete docking pose. Affinity mapping using photoreactive AVG analogs identified the interface of polymerase core, capping, and connector domains as a molecular target site. A first-generation lead showed nanomolar potency against RSV in human airway epithelium organoids but lacked in vivo efficacy. Docking pose-informed synthetic optimization generated orally efficacious AVG-388, which showed potent efficacy in the RSV mouse model when administered therapeutically. This study maps a druggable target in the RSV RdRP and establishes clinical potential of the AVG chemotype against RSV disease.
Collapse
Affiliation(s)
- Julien Sourimant
- Center for Translational Antiviral Research, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Carolin M. Lieber
- Center for Translational Antiviral Research, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Jeong-Joong Yoon
- Center for Translational Antiviral Research, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Mart Toots
- Center for Translational Antiviral Research, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | | | - Venkata Udumula
- Emory Institute for Drug Development, Emory University, Atlanta, GA 30322, USA
| | - Kaori Sakamoto
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Michael G. Natchus
- Emory Institute for Drug Development, Emory University, Atlanta, GA 30322, USA
| | - Joseph Patti
- Aviragen Therapeutics Inc, Alpharetta, GA 30009, USA
| | | | - Richard K. Plemper
- Center for Translational Antiviral Research, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
18
|
Xiang ML, Hu BY, Qi ZH, Wang XN, Xie TZ, Wang ZJ, Ma DY, Zeng Q, Luo XD. Chemistry and bioactivities of natural steroidal alkaloids. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:23. [PMID: 35701630 PMCID: PMC9198197 DOI: 10.1007/s13659-022-00345-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 05/11/2023]
Abstract
Steroidal alkaloids possess the basic steroidal skeleton with a nitrogen atom in rings or side chains incorporated as an integral part of the molecule. They have demonstrated a wide range of biological activities, and some of them have even been developed as therapeutic drugs, such as abiraterone acetate (Zytiga®), a blockbuster drug, which has been used for the treatment of prostate cancer. Structurally diverse natural steroidal alkaloids present a wide spectrum of biological activities, which are attractive for natural product chemistry and medicinal chemistry communities. This review comprehensively covers the structural classification, isolation and various biological activities of 697 natural steroidal alkaloids discovered from 1926 to October 2021, with 363 references being cited.
Collapse
Affiliation(s)
- Mei-Ling Xiang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Bin-Yuan Hu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Zi-Heng Qi
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Xiao-Na Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Tian-Zhen Xie
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Zhao-Jie Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Dan-Yu Ma
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Qi Zeng
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Xiao-Dong Luo
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China.
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| |
Collapse
|
19
|
Zrieq R, Ahmad I, Snoussi M, Noumi E, Iriti M, Algahtani FD, Patel H, Saeed M, Tasleem M, Sulaiman S, Aouadi K, Kadri A. Tomatidine and Patchouli Alcohol as Inhibitors of SARS-CoV-2 Enzymes (3CLpro, PLpro and NSP15) by Molecular Docking and Molecular Dynamics Simulations. Int J Mol Sci 2021; 22:10693. [PMID: 34639036 PMCID: PMC8509278 DOI: 10.3390/ijms221910693] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/24/2022] Open
Abstract
Considering the current dramatic and fatal situation due to the high spreading of SARS-CoV-2 infection, there is an urgent unmet medical need to identify novel and effective approaches for prevention and treatment of Coronavirus disease (COVID 19) by re-evaluating and repurposing of known drugs. For this, tomatidine and patchouli alcohol have been selected as potential drugs for combating the virus. The hit compounds were subsequently docked into the active site and molecular docking analyses revealed that both drugs can bind the active site of SARS-CoV-2 3CLpro, PLpro, NSP15, COX-2 and PLA2 targets with a number of important binding interactions. To further validate the interactions of promising compound tomatidine, Molecular dynamics study of 100 ns was carried out towards 3CLpro, NSP15 and COX-2. This indicated that the protein-ligand complex was stable throughout the simulation period, and minimal backbone fluctuations have ensued in the system. Post dynamic MM-GBSA analysis of molecular dynamics data showed promising mean binding free energy 47.4633 ± 9.28, 51.8064 ± 8.91 and 54.8918 ± 7.55 kcal/mol, respectively. Likewise, in silico ADMET studies of the selected ligands showed excellent pharmacokinetic properties with good absorption, bioavailability and devoid of toxicity. Therefore, patchouli alcohol and especially, tomatidine may provide prospect treatment options against SARS-CoV-2 infection by potentially inhibiting virus duplication though more research is guaranteed and secured.
Collapse
Affiliation(s)
- Rafat Zrieq
- Department of Public Health, College of Public Health and Health Informatics, University of Ha’il, Ha’il 81451, Saudi Arabia; (R.Z.); (F.D.A.)
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405, India; (I.A.); (H.P.)
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha’il City, P.O. 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.S.)
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), University of Monastir, Higher Institute of Biotechnology of Monastir, Avenue Tahar Haddad, BP74, Monastir 5000, Tunisia
| | - Emira Noumi
- Department of Biology, College of Science, University of Ha’il City, P.O. 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.S.)
- Laboratory of Bioresources: Integrative Biology and Valorization, (LR14-ES06), University of Monastir, Higher Institute of Biotechnology of Monastir, Avenue Tahar Haddad, BP74, Monastir 5000, Tunisia
| | - Marcello Iriti
- Department of Agricultural and Environmental Sciences, Università degli Studi di Milano, 20133 Milano, Italy
- Phytochem Lab., Department of Agricultural and Environmental Sciences, Università degli Studi di Milano, 20133 Milano, Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), 50121 Firenze, Italy
- BAT Center—Interuniversity Center for Studies on Bioispired Agro-Environmental Technology, University of Napoli “Federico II”, Portici, 80055 Napoli, Italy
| | - Fahad D. Algahtani
- Department of Public Health, College of Public Health and Health Informatics, University of Ha’il, Ha’il 81451, Saudi Arabia; (R.Z.); (F.D.A.)
- Molecular Diagnostic and Personalized Therapeutics Unit, University of Ha’il, Ha’il 81451, Saudi Arabia
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405, India; (I.A.); (H.P.)
| | - Mohd Saeed
- Department of Biology, College of Science, University of Ha’il City, P.O. 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.S.)
| | - Munazzah Tasleem
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China;
| | - Shadi Sulaiman
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Ha’il, Ha’il 81451, Saudi Arabia;
| | - Kaïss Aouadi
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia;
- Faculty of Science of Monastir, University of Monastir, Avenue of the Environment, Monastir 5019, Tunisia
| | - Adel Kadri
- Department of Chemistry, Faculty of Science and Arts of Baljurashi, Albaha University, Al Bahah 65731, Saudi Arabia;
- Faculty of Science of Sfax, Department of Chemistry, University of Sfax, B.P. 1171, Sfax 3000, Tunisia
| |
Collapse
|
20
|
Dirks ML, Seale JT, Collins JM, McDougal OM. Review: Veratrum californicum Alkaloids. Molecules 2021; 26:5934. [PMID: 34641477 PMCID: PMC8513088 DOI: 10.3390/molecules26195934] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/22/2021] [Accepted: 09/25/2021] [Indexed: 12/02/2022] Open
Abstract
Veratrum spp. grow throughout the world and are especially prevalent in high mountain meadows of North America. All parts of Veratrum plants have been used for the treatment of ailments including injuries, hypertension, and rheumatic pain since as far back as the 1600s. Of the 17-45 Veratrum spp., Veratrum californicum alkaloids have been proven to possess favorable medicinal properties associated with inhibition of hedgehog (Hh) pathway signaling. Aberrant Hh signaling leads to proliferation of over 20 cancers, including basal cell carcinoma, prostate and colon among others. Six of the most well-studied V. californicum alkaloids are cyclopamine (1), veratramine (2), isorubijervine (3), muldamine (4), cycloposine (5), and veratrosine (6). Recent inspection of the ethanolic extract from V. californicum root and rhizome via liquid chromatography-mass spectrometry has detected up to five additional alkaloids that are proposed to be verazine (7), etioline (8), tetrahydrojervine (9), dihydrojervine (10), 22-keto-26-aminocholesterol (11). For each alkaloid identified or proposed in V. californicum, this review surveys literature precedents for extraction methods, isolation, identification, characterization and bioactivity to guide natural product drug discovery associated with this medicinal plant.
Collapse
Affiliation(s)
- Madison L. Dirks
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA; (M.L.D.); (J.T.S.)
| | - Jared T. Seale
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA; (M.L.D.); (J.T.S.)
| | - Joseph M. Collins
- Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID 83725, USA;
| | - Owen M. McDougal
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA; (M.L.D.); (J.T.S.)
| |
Collapse
|
21
|
A condensate-hardening drug blocks RSV replication in vivo. Nature 2021; 595:596-599. [PMID: 34234347 DOI: 10.1038/s41586-021-03703-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/07/2021] [Indexed: 12/31/2022]
Abstract
Biomolecular condensates have emerged as an important subcellular organizing principle1. Replication of many viruses, including human respiratory syncytial virus (RSV), occurs in virus-induced compartments called inclusion bodies (IBs) or viroplasm2,3. IBs of negative-strand RNA viruses were recently shown to be biomolecular condensates that form through phase separation4,5. Here we report that the steroidal alkaloid cyclopamine and its chemical analogue A3E inhibit RSV replication by disorganizing and hardening IB condensates. The actions of cyclopamine and A3E were blocked by a point mutation in the RSV transcription factor M2-1. IB disorganization occurred within minutes, which suggests that these molecules directly act on the liquid properties of the IBs. A3E and cyclopamine inhibit RSV in the lungs of infected mice and are condensate-targeting drug-like small molecules that have in vivo activity. Our data show that condensate-hardening drugs may enable the pharmacological modulation of not only many previously undruggable targets in viral replication but also transcription factors at cancer-driving super-enhancers6.
Collapse
|
22
|
Gao Y, Cao J, Xing P, Altmeyer R, Zhang Y. Evaluation of Small Molecule Combinations against Respiratory Syncytial Virus In Vitro. Molecules 2021; 26:molecules26092607. [PMID: 33946996 PMCID: PMC8125180 DOI: 10.3390/molecules26092607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/03/2021] [Accepted: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major pathogen that causes severe lower respiratory tract infection in infants, the elderly and the immunocompromised worldwide. At present no approved specific drugs or vaccines are available to treat this pathogen. Recently, several promising candidates targeting RSV entry and multiplication steps are under investigation. However, it is possible to lead to drug resistance under the long-term treatment. Therapeutic combinations constitute an alternative to prevent resistance and reduce antiviral doses. Therefore, we tested in vitro two-drug combinations of fusion inhibitors (GS5806, Ziresovir and BMS433771) and RNA-dependent RNA polymerase complex (RdRp) inhibitors (ALS8176, RSV604, and Cyclopamine). The statistical program MacSynergy II was employed to determine synergism, additivity or antagonism between drugs. From the result, we found that combinations of ALS8176 and Ziresovir or GS5806 exhibit additive effects against RSV in vitro, with interaction volume of 50 µM2% and 31 µM2% at 95% confidence interval, respectively. On the other hand, all combinations between fusion inhibitors showed antagonistic effects against RSV in vitro, with volume of antagonism ranging from −50 µM2 % to −176 µM2 % at 95% confidence interval. Over all, our results suggest the potentially therapeutic combinations in combating RSV in vitro could be considered for further animal and clinical evaluations.
Collapse
|
23
|
de Souza Cardoso R, Viana RMM, Vitti BC, Coelho ACL, de Jesus BLS, de Paula Souza J, Pontelli MC, Murakami T, Ventura AM, Ono A, Arruda E. Human Respiratory Syncytial Virus Infection in a Human T Cell Line Is Hampered at Multiple Steps. Viruses 2021; 13:v13020231. [PMID: 33540662 PMCID: PMC7913106 DOI: 10.3390/v13020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 11/19/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is the most frequent cause of severe respiratory disease in children. The main targets of HRSV infection are epithelial cells of the respiratory tract, and the great majority of the studies regarding HRSV infection are done in respiratory cells. Recently, the interest on respiratory virus infection of lymphoid cells has been growing, but details of the interaction of HRSV with lymphoid cells remain unknown. Therefore, this study was done to assess the relationship of HRSV with A3.01 cells, a human CD4+ T cell line. Using flow cytometry and fluorescent focus assay, we found that A3.01 cells are susceptible but virtually not permissive to HRSV infection. Dequenching experiments revealed that the fusion process of HRSV in A3.01 cells was nearly abolished in comparison to HEp-2 cells, an epithelial cell lineage. Quantification of viral RNA by RT-qPCR showed that the replication of HRSV in A3.01 cells was considerably reduced. Western blot and quantitative flow cytometry analyses demonstrated that the production of HRSV proteins in A3.01 was significantly lower than in HEp-2 cells. Additionally, using fluorescence in situ hybridization, we found that the inclusion body-associated granules (IBAGs) were almost absent in HRSV inclusion bodies in A3.01 cells. We also assessed the intracellular trafficking of HRSV proteins and found that HRSV proteins colocalized partially with the secretory pathway in A3.01 cells, but these HRSV proteins and viral filaments were present only scarcely at the plasma membrane. HRSV infection of A3.01 CD4+ T cells is virtually unproductive as compared to HEp-2 cells, as a result of defects at several steps of the viral cycle: Fusion, genome replication, formation of inclusion bodies, recruitment of cellular proteins, virus assembly, and budding.
Collapse
Affiliation(s)
- Ricardo de Souza Cardoso
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (T.M.); (A.O.)
| | - Rosa Maria Mendes Viana
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Brenda Cristina Vitti
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Ana Carolina Lunardello Coelho
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Bruna Laís Santos de Jesus
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Juliano de Paula Souza
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Marjorie Cornejo Pontelli
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Tomoyuki Murakami
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (T.M.); (A.O.)
| | - Armando Morais Ventura
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil;
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (T.M.); (A.O.)
| | - Eurico Arruda
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
- Correspondence:
| |
Collapse
|
24
|
Hu M, Bogoyevitch MA, Jans DA. Impact of Respiratory Syncytial Virus Infection on Host Functions: Implications for Antiviral Strategies. Physiol Rev 2020; 100:1527-1594. [PMID: 32216549 DOI: 10.1152/physrev.00030.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of viral respiratory tract infection in infants, the elderly, and the immunocompromised worldwide, causing more deaths each year than influenza. Years of research into RSV since its discovery over 60 yr ago have elucidated detailed mechanisms of the host-pathogen interface. RSV infection elicits widespread transcriptomic and proteomic changes, which both mediate the host innate and adaptive immune responses to infection, and reflect RSV's ability to circumvent the host stress responses, including stress granule formation, endoplasmic reticulum stress, oxidative stress, and programmed cell death. The combination of these events can severely impact on human lungs, resulting in airway remodeling and pathophysiology. The RSV membrane envelope glycoproteins (fusion F and attachment G), matrix (M) and nonstructural (NS) 1 and 2 proteins play key roles in modulating host cell functions to promote the infectious cycle. This review presents a comprehensive overview of how RSV impacts the host response to infection and how detailed knowledge of the mechanisms thereof can inform the development of new approaches to develop RSV vaccines and therapeutics.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Blockus S, Sake SM, Wetzke M, Grethe C, Graalmann T, Pils M, Le Goffic R, Galloux M, Prochnow H, Rox K, Hüttel S, Rupcic Z, Wiegmann B, Dijkman R, Rameix-Welti MA, Eléouët JF, Duprex WP, Thiel V, Hansen G, Brönstrup M, Haid S, Pietschmann T. Labyrinthopeptins as virolytic inhibitors of respiratory syncytial virus cell entry. Antiviral Res 2020; 177:104774. [PMID: 32197980 DOI: 10.1016/j.antiviral.2020.104774] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/03/2020] [Accepted: 03/12/2020] [Indexed: 10/25/2022]
Abstract
Acute lower respiratory tract infections (ALRI) caused by respiratory syncytial virus (RSV) are associated with a severe disease burden among infants and elderly patients. Treatment options are limited. While numerous drug candidates with different viral targets are under development, the utility of RSV entry inhibitors is challenged by a low resistance barrier and by single mutations causing cross-resistance against a wide spectrum of fusion inhibitor chemotypes. We developed a cell-based screening assay for discovery of compounds inhibiting infection with primary RSV isolates. Using this system, we identified labyrinthopeptin A1 and A2 (Laby A1/A2), lantibiotics isolated from Actinomadura namibiensis, as effective RSV cell entry inhibitors with IC50s of 0.39 μM and 4.97 μM, respectively, and with favourable therapeutic index (>200 and > 20, respectively). Both molecules were active against multiple RSV strains including primary isolates and their antiviral activity against RSV was confirmed in primary human airway cells ex vivo and a murine model in vivo. Laby A1/A2 were antiviral in prophylactic and therapeutic treatment regimens and displayed synergistic activity when applied in combination with each other. Mechanistic studies showed that Laby A1/A2 exert virolytic activity likely by binding to phosphatidylethanolamine moieties within the viral membrane and by disrupting virus particle membrane integrity. Probably due to its specific mode of action, Laby A1/A2 antiviral activity was not affected by common resistance mutations to known RSV entry inhibitors. Taken together, Laby A1/A2 represent promising candidates for development as RSV inhibitors. Moreover, the cell-based screening system with primary RSV isolates described here should be useful to identify further antiviral agents.
Collapse
Affiliation(s)
- Sebastian Blockus
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Svenja M Sake
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Martin Wetzke
- Department for Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany; German Centre of Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Christina Grethe
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Theresa Graalmann
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany; Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Marina Pils
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Ronan Le Goffic
- Unité de Virologie et Immunologie Moléculaires, INRA, Université Paris Saclay, Jouy-en-Josas, France
| | - Marie Galloux
- Unité de Virologie et Immunologie Moléculaires, INRA, Université Paris Saclay, Jouy-en-Josas, France
| | - Hans Prochnow
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; German Centre of Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; German Centre of Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Stephan Hüttel
- Department Microbial Drugs, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Zeljka Rupcic
- Department Microbial Drugs, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Bettina Wiegmann
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany; Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Ronald Dijkman
- Institute of Virology and Immunology (IVI), Bern and Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Switzerland; Institute for Infectious Diseases, University of Bern, Switzerland
| | - Marie-Anne Rameix-Welti
- UMR1173, Institute National de la Santé et de la Recherche Médicale (INSERM), Université de Versailles St. Quentin, Montigny-le-Bretonneux, France
| | - Jean-François Eléouët
- Unité de Virologie et Immunologie Moléculaires, INRA, Université Paris Saclay, Jouy-en-Josas, France
| | - W Paul Duprex
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Volker Thiel
- Institute of Virology and Immunology (IVI), Bern and Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Gesine Hansen
- Department for Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; German Centre of Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Sibylle Haid
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.
| | - Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany; German Centre of Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
26
|
Norris MJ, Malhi M, Duan W, Ouyang H, Granados A, Cen Y, Tseng YC, Gubbay J, Maynes J, Moraes TJ. Targeting Intracellular Ion Homeostasis for the Control of Respiratory Syncytial Virus. Am J Respir Cell Mol Biol 2019; 59:733-744. [PMID: 30095982 DOI: 10.1165/rcmb.2017-0345oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of mortality in infants and young children. Despite the RSV disease burden, no vaccine is available, and treatment remains nonspecific. New drug candidates are needed to combat RSV. Toward this goal, we screened over 2,000 compounds to identify approved drugs with novel anti-RSV activity. Cardiac glycosides, inhibitors of the membrane-bound Na+/K+-ATPase, were identified to have anti-RSV activity. Cardiac glycosides diminished RSV infection in human epithelial type 2 cells and in primary human airway epithelial cells grown at an air-liquid interface. Digoxin, a U.S. Food and Drug Administration-approved cardiac glycoside, was also able to inhibit infection of primary nasal epithelial cells with community isolates of RSV. Our results suggest that the antiviral effects of cardiac glycosides may be dependent on changes in the intracellular Na+ and K+ composition. Consistent with this mechanism, we demonstrated that the ionophoric antibiotics salinomycin, valinomycin, and monensin inhibited RSV in human epithelial type 2 cells and primary nasal epithelial cells. Our data indicate that the K+/Na+-sensitive steps in the RSV life cycle occur within the initial 4 hours of viral infection but do not include virus binding/entry. Rather, our findings demonstrated a negative effect on the RSV transcription and/or replication process. Overall, this work suggests that targeting intracellular ion concentrations offers a novel antiviral strategy.
Collapse
Affiliation(s)
- Michael J Norris
- 1 Department of Laboratory Medicine and Pathobiology and.,2 Program in Translational Medicine
| | - Manpreet Malhi
- 3 Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,4 Program in Molecular Medicine
| | | | | | - Andrea Granados
- 1 Department of Laboratory Medicine and Pathobiology and.,5 Public Health Ontario, Toronto, Ontario, Canada
| | | | | | | | - Jason Maynes
- 4 Program in Molecular Medicine.,6 Department of Anesthesia and Pain Medicine, and
| | - Theo J Moraes
- 1 Department of Laboratory Medicine and Pathobiology and.,2 Program in Translational Medicine.,7 Division of Respiratory Medicine, Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada; and
| |
Collapse
|
27
|
Roumana A, Yektaoğlu A, Pliatsika D, Bantzi M, Nikolaropoulos SS, Giannis A, Fousteris MA. New Spiro-Lactam C- nor
- D
- homo
Steroids. European J Org Chem 2018. [DOI: 10.1002/ejoc.201800667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Aggeliki Roumana
- Laboratory of Medicinal Chemistry; Department of Pharmacy; University of Patras; -26500 Patras GR Greece
| | - Aybike Yektaoğlu
- Institute of Organic Chemistry; Faculty of Chemistry and Mineralogy; University of Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Dimanthi Pliatsika
- Laboratory of Medicinal Chemistry; Department of Pharmacy; University of Patras; -26500 Patras GR Greece
- Institute of Organic Chemistry; Faculty of Chemistry and Mineralogy; University of Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Marina Bantzi
- Institute of Organic Chemistry; Faculty of Chemistry and Mineralogy; University of Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Sotiris S. Nikolaropoulos
- Laboratory of Medicinal Chemistry; Department of Pharmacy; University of Patras; -26500 Patras GR Greece
| | - Athanassios Giannis
- Institute of Organic Chemistry; Faculty of Chemistry and Mineralogy; University of Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Manolis A. Fousteris
- Laboratory of Medicinal Chemistry; Department of Pharmacy; University of Patras; -26500 Patras GR Greece
| |
Collapse
|
28
|
Mucke HA. Patent highlights August-September 2017. Pharm Pat Anal 2018; 7:7-14. [PMID: 29219751 DOI: 10.4155/ppa-2017-0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/07/2017] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
29
|
Rincheval V, Lelek M, Gault E, Bouillier C, Sitterlin D, Blouquit-Laye S, Galloux M, Zimmer C, Eleouet JF, Rameix-Welti MA. Functional organization of cytoplasmic inclusion bodies in cells infected by respiratory syncytial virus. Nat Commun 2017; 8:563. [PMID: 28916773 PMCID: PMC5601476 DOI: 10.1038/s41467-017-00655-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/17/2017] [Indexed: 12/23/2022] Open
Abstract
Infection of cells by respiratory syncytial virus induces the formation of cytoplasmic inclusion bodies (IBs) where all the components of the viral RNA polymerase complex are concentrated. However, the exact organization and function of these IBs remain unclear. In this study, we use conventional and super-resolution imaging to dissect the internal structure of IBs. We observe that newly synthetized viral mRNA and the viral transcription anti-terminator M2-1 concentrate in IB sub-compartments, which we term “IB-associated granules” (IBAGs). In contrast, viral genomic RNA, the nucleoprotein, the L polymerase and its cofactor P are excluded from IBAGs. Live imaging reveals that IBAGs are highly dynamic structures. Our data show that IBs are the main site of viral RNA synthesis. They further suggest that shortly after synthesis in IBs, viral mRNAs and M2-1 transiently concentrate in IBAGs before reaching the cytosol and suggest a novel post-transcriptional function for M2-1. Respiratory syncytial virus (RSV) induces formation of inclusion bodies (IBs) sheltering viral RNA synthesis. Here, Rincheval et al. identify highly dynamic IB-associated granules (IBAGs) that accumulate newly synthetized viral mRNA and the viral M2-1 protein but exclude viral genomic RNA and RNA polymerase complexes.
Collapse
Affiliation(s)
- Vincent Rincheval
- UMR1173, INSERM, Université de Versailles St. Quentin, Montigny le Bretonneux, 78180, France
| | - Mickael Lelek
- Institut Pasteur Unité Imagerie et Modélisation, CNRS UMR 3691; C3BI, USR 3756, IP CNRS, Paris, 75015, France
| | - Elyanne Gault
- UMR1173, INSERM, Université de Versailles St. Quentin, Montigny le Bretonneux, 78180, France.,AP-HP, Laboratoire de Microbiologie, Hôpital Ambroise Paré, Boulogne-Billancourt, 92104, France
| | - Camille Bouillier
- UMR1173, INSERM, Université de Versailles St. Quentin, Montigny le Bretonneux, 78180, France
| | - Delphine Sitterlin
- UMR1173, INSERM, Université de Versailles St. Quentin, Montigny le Bretonneux, 78180, France
| | - Sabine Blouquit-Laye
- UMR1173, INSERM, Université de Versailles St. Quentin, Montigny le Bretonneux, 78180, France
| | - Marie Galloux
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Université Paris-Saclay, Jouy-en-Josas, 78352, France
| | - Christophe Zimmer
- Institut Pasteur Unité Imagerie et Modélisation, CNRS UMR 3691; C3BI, USR 3756, IP CNRS, Paris, 75015, France
| | - Jean-François Eleouet
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Université Paris-Saclay, Jouy-en-Josas, 78352, France
| | - Marie-Anne Rameix-Welti
- UMR1173, INSERM, Université de Versailles St. Quentin, Montigny le Bretonneux, 78180, France. .,AP-HP, Laboratoire de Microbiologie, Hôpital Ambroise Paré, Boulogne-Billancourt, 92104, France.
| |
Collapse
|
30
|
Krieger J, Smeilus T, Schackow O, Giannis A. Lewis Acid Mediated Nazarov Cyclization as a Convergent and Enantioselective Entry to C
-nor-D
-homo-Steroids. Chemistry 2017; 23:5000-5004. [DOI: 10.1002/chem.201701008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Johannes Krieger
- Institut für Organische Chemie; Fakultät für Chemie und Mineralogie; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Toni Smeilus
- Institut für Organische Chemie; Fakultät für Chemie und Mineralogie; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Oliver Schackow
- Institut für Organische Chemie; Fakultät für Chemie und Mineralogie; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Athanassios Giannis
- Institut für Organische Chemie; Fakultät für Chemie und Mineralogie; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| |
Collapse
|
31
|
Shook BC, Lin K. Recent Advances in Developing Antiviral Therapies for Respiratory Syncytial Virus. Top Curr Chem (Cham) 2017; 375:40. [DOI: 10.1007/s41061-017-0129-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/01/2017] [Indexed: 01/23/2023]
|
32
|
Drug candidates and model systems in respiratory syncytial virus antiviral drug discovery. Biochem Pharmacol 2017; 127:1-12. [DOI: 10.1016/j.bcp.2016.09.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/16/2016] [Indexed: 12/11/2022]
|